You are on page 1of 10

OncoImmunology

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/koni20

Combination checkpoint therapy with anti-PD-1


and anti-BTLA results in a synergistic therapeutic
effect against murine glioblastoma

John Choi, Ravi Medikonda, Laura Saleh, Timothy Kim, Ayush Pant,
Siddhartha Srivastava, Young-Hoon Kim, Christina Jackson, Luqing Tong,
Denis Routkevitch, Christopher Jackson, Dimitrios Mathios, Tianna Zhao,
Hyerim Cho, Henry Brem & Michael Lim

To cite this article: John Choi, Ravi Medikonda, Laura Saleh, Timothy Kim, Ayush Pant,
Siddhartha Srivastava, Young-Hoon Kim, Christina Jackson, Luqing Tong, Denis Routkevitch,
Christopher Jackson, Dimitrios Mathios, Tianna Zhao, Hyerim Cho, Henry Brem & Michael Lim
(2021) Combination checkpoint therapy with anti-PD-1 and anti-BTLA results in a synergistic
therapeutic effect against murine glioblastoma, OncoImmunology, 10:1, 1956142, DOI:
10.1080/2162402X.2021.1956142

To link to this article: https://doi.org/10.1080/2162402X.2021.1956142

© 2021 The Author(s). Published with View supplementary material


license by Taylor & Francis Group, LLC.

Published online: 29 Aug 2021. Submit your article to this journal

Article views: 2578 View related articles

View Crossmark data Citing articles: 7 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=koni20
ONCOIMMUNOLOGY
2021, VOL. 10, NO. 1, e1956142 (9 pages)
https://doi.org/10.1080/2162402X.2021.1956142

ORIGINAL RESEARCH

Combination checkpoint therapy with anti-PD-1 and anti-BTLA results in a synergistic


therapeutic effect against murine glioblastoma
John Choia*, Ravi Medikondaa*, Laura Saleha, Timothy Kima, Ayush Panta, Siddhartha Srivastavaa, Young-Hoon Kimb,
Christina Jacksona, Luqing Tonga, Denis Routkevitcha, Christopher Jacksona, Dimitrios Mathios a, Tianna Zhaoa,
Hyerim Choc, Henry Brema, and Michael Lim a
a
Department of Neurosurgery, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, USA; bDepartment of Neurosurgery, College of
Medicine, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea; cDepartment of Radiology, Seoul National University College of
Medicine, Seoul, Republic of Korea

ABSTRACT ARTICLE HISTORY


Clinical trials involving anti-programmed cell death protein-1 (anti-PD-1) failed to demonstrate improved Received 25 January 2021
overall survival in glioblastoma (GBM) patients. This may be due to the expression of alternative check­ Revised 12 July 2021
points such as B- and T- lymphocyte attenuator (BTLA) on several immune cell types including regulatory Accepted 12 July 2021
T cells. Murine GBM models indicate that there is significant upregulation of BTLA in the tumor micro­ KEYWORDS
environment (TME) with associated T cell exhaustion. We investigate the use of antibodies against BTLA glioblastoma
and PD-1 on reversing immunosuppression and increasing long-term survival in a murine GBM model. immunotherapy; anti-PD-1;
C57BL/6 J mice were implanted with the murine glioma cell line GL261 and randomized into 4 arms: (i) anti-BTLA; B and
control, (ii) anti-PD-1, (iii) anti-BTLA, and (iv) anti-PD-1 + anti-BTLA. Kaplan–Meier curves were generated T lymphocyte attenuator;
for all arms. Flow cytometric analysis of blood and brains were done on days 11 and 16 post-tumor immune checkpoint inhibitor
implantation. Tumor-bearing mice treated with a combination of anti-PD-1 and anti-BTLA therapy therapy; glioblastoma
experienced improved overall long-term survival (60%) compared to anti-PD-1 (20%) or anti-BTLA (0%)
alone (P = .003). Compared to monotherapy with anti-PD-1, mice treated with combination therapy also
demonstrated increased expression of CD4+ IFN-γ (P < .0001) and CD8+ IFN-γ (P = .0365), as well as
decreased levels of CD4+ FoxP3+ regulatory T cells on day 16 in the brain (P = .0136). This is the first
preclinical investigation into the effects of combination checkpoint blockade with anti-PD-1 and anti-BTLA
treatment in GBM. We also show a direct effect on activated immune cell populations such as CD4+ and
CD8 + T cells and immunosuppressive regulatory T cells through this combination therapy.

Introduction (TME).8,9 Moreover, regulatory T cells (Tregs) – an immuno­


suppressive T cell population usually not seen in the brain –
Glioblastoma (GBM) is a highly aggressive primary brain
have significantly increased representation within high-grade
tumor with a dismal prognosis.1 Despite current standards of
gliomas such as GBM.10
care involving maximal surgical resection, chemotherapy, and
With anti-PD-1 monotherapy failing to show significant
radiation therapy, median overall survival (mOS) is approxi­
clinical benefit, several studies have examined the efficacy of
mately 19 months.2,3 Recently, the advent of immune check­
combining multiple checkpoint inhibitors in order to target
point blockade (ICB) with antibodies against programmed cell
different pathways of immunosuppression in GBM.11,12 B and
death protein-1 (anti-PD-1) served as a novel immunotherapy
T lymphocyte attenuator (BTLA) is a co-inhibitory checkpoint
treatment strategy that has shown promise in many solid
molecule that is structurally related to PD-1.13 Previous studies
tumors including melanoma and non-small cell lung cancer
have shown that BTLA is highly expressed on tumor-specific
(NSCLC).4,5
T cells in cancer patients14 and is upregulated in several cancers
Given the success of ICB in other cancers, there has been
including gastric cancer,15 hepatocellular carcinoma,16 and
avid interest in applying anti-PD-1 therapy to GBM. However,
GBM.17 Furthermore, it has been shown that increased BTLA
the recent phase III CheckMate 143 clinical trial evaluating
expression correlates with resistance to anti-PD-1 immu­
anti-PD-1 in patients with GBM failed to improve mOS com­
notherapy and poor clinical outcomes.15,16
pared to current standard of care.6,7 The lack of significant
In the context of GBM, the ligand for BTLA – Herpes virus
clinical benefit from anti-PD-1 therapy is thought to be in part
entry mediator (HVEM) – is overexpressed on high-grade
due to both intrinsic resistance to the immune system due to
gliomas and is associated with the suppression of T cell-
poor antigen presentation/priming, low-quality neoantigens,
mediated anti-tumor activity in the GBM TME.18 Given the
and relatively low mutational burden, as well as extrinsic resis­
likely immunosuppressive effect of BTLA in the GBM TME, we
tance from an immunosuppressive tumor microenvironment
sought to evaluate the survival efficacy of anti-BTLA therapy in

CONTACT Michael Lim mklim@stanford.edu Department of Neurosurgery, Johns Hopkins University, Baltimore 21231, USA
*Indicates authors contributed equally to the manuscript
Supplemental data for this article can be accessed on the publisher’s website.
© 2021 The Author(s). Published with license by Taylor & Francis Group, LLC.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (http://creativecommons.org/licenses/by-nc/4.0/), which permits
unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
e1956142-2 J. CHOI ET AL.

combination with anti-PD-1 in a murine GBM model and assays. 120 μl blood was collected retro-orbitally from mice
assess the immunological effects of this dual immune check­ under deep anesthesia on days 11 and 16 according to our
point blockade therapy. IACUC protocol. Red blood cells were lysed using ACK lysis
buffer (ThermoFisher) and resuspended in PBS for further
cytometric analysis. Brains were removed, tissue was mechani­
Materials and methods cally dissociated through a 70 μm filter, and homogenates were
Murine glioma model and cell lines centrifuged (ThermoFisher Sorvall Legend X1R centrifuge) in
a 30%/70% Percoll® (Sigma-Aldrich) gradient at 2200 rpm for
6–8 week-old C57BL/6 J wild-type female mice were main­ 20 minutes without brakes to separate out brain myeloid cells
tained at the Johns Hopkins University Animal Facility per the and lymphocytes from tumor cells and myelin. Brain immune
Institutional Animal Care and Use Committee (IACUC) pro­ cells were extracted at the 30%/70% interface and resuspended
tocol. For all experiments, mice were anesthetized with in phosphate-buffered saline (PBS) buffer (Sigma-Aldrich) for
Ketathesia (100 mg/kg)/xylazine (10 mg/kg) by intra- further cytometric analysis.
peritoneal (i.p.) injection and had topical eye gel for lubrica­
tion. After every procedure, mice were placed on a heating pad
and observed until fully recovered. Orthotopic murine glioma Flow cytometric analysis of murine immune cells
models utilized GL261-Luc2 (RRID:CVCL_X986) cells grown Mouse immune cells were stained for Live/Dead (L/D)
in DMEM (Life Technologies) + 10% FBS (Sigma-Aldrich) + (Invitrogen), CD45, CD3, CD4, CD8, PD-1, IFN-γ, FoxP3,
1% penicillin-streptomycin (Life Technologies) as described in and BTLA (Supplementary Figure S1 for antibody fluoro­
previous studies (Zeng, 2013). GL261-Luc2 cells were obtained phores). To stain for the intracellular marker IFN-γ and intra­
from Reardon Lab, Dana Farber Cancer Institute. All experi­ nuclear marker FoxP3, samples were fixed in 1:3 fixation/
ments were performed with mycoplasma-free cells. 1.3e5 permeabilization concentrate:diluent mixture (eBioscence) for
GL261-Luc2 cells in a volume of 2 μl were stereotactically 30 minutes and subsequently stained in permeabilization buf­
injected 2 mm posterior to the coronal suture, 2 mm lateral fer (eBioscience). Fluorescence minus one (FMO) was used to
to the sagittal suture, and 3 mm deep to the cortical surface in control for data spread due to multiple fluorochromes and
the area of the left striatum. After implantation of tumor cells, nonbinary expression of markers such as IFN-γ. Flow data
mice were assessed for tumor growth on post-implantation day was acquired using a FACSCelesta flow cytometer (BD) and
7 using bioluminescent IVIS imaging (PerkinElmer). analyzed using FlowJo (BD). Nonviable cells and doublets were
Mice with tumor burden at day 7 were then randomly excluded by forward versus side scatter gating, forward scatter
separated into control (non-treated) and treatment arms. height versus forward scatter area gating, and L/D staining.
Survival experiments were repeated in triplicate with 8–10
mice in each control or treatment arm. Animals were eutha­
nized according to humane endpoints, including central ner­ Co-culture and ELISA
vous system disturbances, hunched posture, lethargy, weight For IFN-γ assays, 5e3 CD45.2+ CD3+ CD8 + T cells were
loss, and inability to ambulate per our IACUC protocol. Long- sorted from each mouse brain and co-cultured with 100 μg/
term survivors were rechallenged with re-implantation of ml GL261-Luc2 tumor cell lysate and 25e3 dendritic cells iso­
GL261-Luc2 in the right striatum on post-implantation day lated from CD45.1 mouse spleen (isolated with a pan-dendritic
60. Additionally, 8 naïve control mice were implanted with cell isolation kit) (Miltenyi Biotec) in a 96-well round bot­
tumor the day of rechallenge to serve as controls. tomed plate with T cell media (RPMI 1640 + 10% FBS + 1%
NEAA + 1% 2-Mercaptoethanol + 1% Penicillin/
Therapeutic antibodies Streptomycin). Co-cultured cells were incubated at 37°C for
48 hours. Supernatant was collected for subsequent ELISA for
G4 hybridomas were cultured and used to develop hamster IFN-γ and run on a plate reader (PerkinElmer Victor3 1420
monoclonal antibodies (mAbs) against murine PD-1, as Multilabel plate reader).
described in previous studies (Hirano F, 2005). Individual
treatment dose was 200 μg per animal on post-implantation
days 10, 12, and 14 for anti-PD-1 monotherapy and combina­ Depletion study
tion therapy arms. Anti-murine BTLA antibody was received GL261-luc2 tumor was confirmed in mice on post-
from Bristol Myers Squibb (BMS) and stored at −80°C in 1 mg/ implantation day 7. After randomization, tumor bearing mice
mL aliquots. Per the manufacturer, anti-BTLA is a non- received IP injections of either anti-CD4 (clone GK1.5, Bio
depleting, blocking antibody with a defective Fc region pre­ X Cell, catalog#: BE0003-1) at 200 μg/dose or anti-CD8
venting antibody-dependent cellular cytotoxicity. This anti­ (clone 2.43, Bio X Cell, catalog#: BE0004-1) at 500 μg/dose.
body was administered in three 400 μg doses on post- Control mice received 200 μL of PBS IP. All depletion anti­
implantation days 7, 10, and 14. bodies were administered 48 hours and 24 hours prior to the
administration of the first dose of anti-BTLA treatment.
Depletion antibodies were administered every seven days
Immune cell harvest and isolation
thereafter until conclusion of the study. T cell depletion was
Mice were deeply anesthetized or euthanized before harvesting confirmed via flow cytometry of blood in each treatment arm
blood (120 μl, retro-orbital) and brains for immunological on day 14 post-implantation.
ONCOIMMUNOLOGY e1956142-3

Statistical analysis expression on CD3+ CD8 + T cells did not significantly change
from 12.9% on day 7 to 17.1% on day 21 (P = .49) (Figure 1c).
All replicates were biological replicates. Survival was analyzed
Next, expression of BTLA and its ligand HVEM was studied
via Kaplan-Meier method and compared by log-rank (Mantel-
in the glioma setting on various tumor-infiltrating immune cell
Cox) test. Calculated variables were treated as continuous
populations including CD45+ CD3 + T cells, CD45+ CD11b+
variables under the assumption that data follow Student
cells (commonly denoting macrophages), CD45+ CD11 c
T-distribution. Mouse experimental data were analyzed using
+ cells (commonly denoting macrophages and dendritic cells)
a 2-tailed Student’s T-test for experiments containing 2 groups.
and CD45+ CD19 + B cells (Supplemental Figure S1). There
All data were analyzed using GraphPad Prism 8 and values of
was significantly lower HVEM expression on CD3+ TILs com­
P < .05 were considered statistically significant.
pared to tumor-infiltrating CD11b+ cells (P < .0001), CD11 c
+ cells (P < .0001), or CD19 + B cells (P < .0001) (Supplemental
Figure S1b). There was significantly lower BTLA expression on
Results CD11 c+ cells compared to CD3 + T cells (P = .0405) and
BTLA expression increases over time on tumor-infiltrating tumor-infiltrating CD11b+ cells (P = .0062) (Supplemental
non-Treg CD4 + T cells Figure S1b). HVEM expression was also analyzed after admin­
istration of control (no treatment), anti-PD-1 alone, anti-BTLA
To evaluate the expression of BTLA over time on immune cells, alone or combination therapy. There was no significant differ­
a time point experiment was conducted in mice on post- ence in HVEM expression on tumor-infiltrating CD11b+ cells,
implantation day 7, 16, and 21 (Figure 1). 1.3e5 GL261-Luc+ CD11 c+ cells, or CD19 + B cells with the four treatment arms.
cells were implanted in the left striatum on day 0. Blood and There was significantly higher HVEM expression on CD3 + T
brain were harvested at each of the three time points for 5 mice cells with anti-BTLA (P = .0135) or combination therapy
after confirming presence of tumor on IVIS. Single cell suspen­ (P = .0285) compared to control (Supplemental Figure S1c).
sions were prepared and stained for flow cytometric analysis of Combination anti-BTLA and anti-PD-1 treatment increases
BTLA. In the blood, BTLA expression significantly increased activation of CD4+ and CD8 + T cells and modulates presence of
on non-Treg CD4 + T cells from 6.1% on day 7 to 55.1% on day Tregs in the brain and blood
16 (P < .0001) and 67.8% on day 21 (P < .0001) (Figure 1b). In In order to assess the immunologic effects of anti-PD-1 and
the brain, BTLA expression on tumor-infiltrating Treg (CD3 anti-BTLA in murine GBM models, blood was harvested on
+ CD4+ FoxP3+) cells did not significantly change from an days 11 and 16 with brains also harvested on day 16. T cells
average of 13.4% on day 7 to 16.2% on day 16 (P = .003) and were gated on forward versus side scatter for lymphocyte
20.0% on day 21 (P = .003) (Figure 1c). However, BTLA populations with exclusion of doublets, which were then
expression on tumor-infiltrating non-Treg (CD3+ CD4 gated for live CD45+ CD3 + T cells and separated into CD4
+ FoxP3-) T cells significantly increased from a mean of + and CD8+ populations (Supplemental Figure S2).
10.1% on day 7 to 37.7% on day 16 (P < . 0001) and 37.8% On day 16 in the brain, IFN-γ expression of CD4+ and CD8
on day 21 (P < .0001) (Figure 1c). On CD3+ CD8 + T cells in + populations were compared between experimental arms,
the blood, BTLA expression increased from 8.2% on day 7 to with 32.9% of CD4+ tumor-infiltrating lymphocytes (TILs)
35.3% on day 21 (P < .001) (Figure 1b). In the brain, BTLA expressing IFN-γ in the combination anti-PD-1 and anti-

Figure 1. Timing of BTLA expression in tumor-bearing mice on days 7, 16, and 21. (a) Representative flow cytometry plots of TILs demonstrating BTLA expression
on day 7, 16, and 21 on CD4+ FoxP3 + T cells, CD4+ FoxP3- T cells, and CD8 + T cells (b)Aggregate (n = 5) flow cytometry data of BTLA expression in PBMCs from
untreated tumor-bearing mice on days 7, 16, and 21. (c) Aggregate (n = 5) flow cytometry data of BTLA expression in TILs of untreated tumor-bearing mice on days 7,
16, and 21. All plots represent mean with standard deviation. Comparison between two groups is made via the Student’s t test. (* p < .05).
e1956142-4 J. CHOI ET AL.

BTLA arm compared to 11.7% in the non-treated (control) therapy (38.7%) compared to control (29.0%, P = .016), anti-
arm, 16.7% in the anti-PD-1 monotherapy arm (P < .0001), and PD-1 monotherapy (21.2%, P = .0016), anti-BTLA monother­
19.1% in the anti-BTLA arm (P = .0033) (Figure 2a, b). apy (18.1%, P = .0063) (Figure 2c). There was no significant
Similarly, 76.3% of CD8+ TILs expressed IFN-γ in the combi­ difference in LAG-3 or TIM-3 expression between control,
nation anti-PD-1 and anti-BTLA arm compared to 43.8%, monotherapy, or combination therapy.
59.9%, and 44.8% in control, anti-PD-1 monotherapy We also studied if anti-PD-1 and anti-BTLA combination
(P = .0365), and anti-BTLA (P = .0004) monotherapy arms, therapy was affecting tumor-infiltrating myeloid or B cell
respectively (Figure 2b). populations. After treating mice with control, anti-PD-1 only,
Of note, in the combination therapy arm, there was anti-BTLA only, or combination therapy, we found no signifi­
a marked decrease in the proportion of CD4+ FoxP3 + T regs cant change in tumor infiltration of CD45+ CD11b+ macro­
of the parent CD4+ TIL population in the brain compared to phages between the four treatment arms. Of note, there was
monotherapy and control arms, with 11.7% of CD4+ TILs a trend toward significantly greater CD45+ CD11c+ macro­
expressing FoxP3 in mice treated with both anti-PD-1 and phage and dendritic cell infiltration of the tumor in the com­
anti-BTLA compared to 36.0% of CD4 + T cells in non- bination therapy arm compared to control (P = .09). Likewise,
treated mice expressing FoxP3 and 27.9% of CD4+ TILs in there was a trend toward significantly greater CD45+ CD19 + B
mice treated with anti-PD-1 monotherapy expressing FoxP3 cell tumor infiltration with combination therapy compared to
(P = .0136) (Figure 2b). However, there was no statistically control (P = .06) (Supplemental Figure S3).
significant difference between combination and anti-BTLA To assess the characteristics of possible homing and periph­
monotherapy arms for FoxP3 expression in the brain, with eral proliferation of lymphocytes, blood was analyzed on day
15.8% of CD4+ TILs in mice with anti-BTLA monotherapy 11, one day after exposure to both anti-PD-1 and anti-BTLA, as
expressing FoxP3 (P = .2349). However, there was a statistically well as on day 16 – the day of brain harvest. Indications of
significant difference in FoxP3 expression between anti-PD-1 T cell homing were present across all arms for CD8 + T cells,
and anti-BTLA monotherapy arms (P = .0483) (Figure 2a, b). with marked decrease in percentage of CD8+ IFN-γ-secreting
Next, we evaluated for the expression of additional co- T cells on day 16 compared to day 11. Furthermore, combina­
stimulatory molecules such as 4–1BB and co-inhibitory mole­ tion treatment with anti-PD-1 and anti-BTLA showed the
cules such as TIM-3 and LAG-3 on TILs. 4–1BB expression on greatest levels of CD4+ and CD8+ IFN-γ expression on day
CD4+ TILs was significantly higher with combination therapy 11 with 21.5% of CD4+ cells and 37.9% of CD8+ cells expres­
(31.6%) compared to control (23. 2%, P = .0098), anti-PD-1 sing IFN-γ (Supplemental Figure S4). Notably, tumor bearing
monotherapy (23.7%, P = .0135), anti-BTLA monotherapy mice treated with combination therapy did not observe
(19.1%, P = .0007) (Figure 2c). Likewise, 4–1BB expression a similar trend in peripheral regulatory T cell distribution on
on CD8+ TILs was significantly higher with combination days 11 and 16; while other monotherapy arms showed

Figure 2. Flow cytometric analysis of T cell populations in mouse brain at day 16 post-tumor implantation. (a) Flow plots demonstrating CD4 and CD8 IFN-γ
expression, 4–1BB expression, and CD4 Foxp3 presence in the brain. (b) Aggregate (n = 5 per group) data of control (no treatment), anti-PD-1, anti-BTLA, and
combination therapy groups for CD4 and CD8 IFN-γ and CD4 Foxp3 presence in the brain. Differences between two treatment arms were analyzed via Student’s t test.
(**** P < .0001, *** P < .001, ** P < .01, * P < .05).
ONCOIMMUNOLOGY e1956142-5

a marked decrease in Tregs between days 11 and 16, mice contralaterally rechallenged/implanted with intracranial
treated with combination therapy showed similar proportions GL261-Luc2. Long-term survivors rechallenged with glioma
of Tregs in the blood on both days (1.52% and 1.56% on days demonstrated significantly better survival than control mice
11 and 16, respectively) (P = .8271) (Figure S3). (P = .0031) (Supplemental Figure S5).
Treatment with anti-BTLA and anti-PD-1 correlate with Depletion of CD4 + T cells or CD8 + T cells diminishes the
increased IFN-γ secretion efficacy of anti-PD-1 and anti-BTLA combination therapy
To further determine if combination therapy with anti-PD The results thus far suggest that anti-PD-1 and anti-BTLA
-1 and anti-BTLA resulted in decreased immunosuppression, combination therapy provide a synergistic survival benefit in
TILs belonging to mice ± treatment with anti-PD-1 monother­ murine glioma (Figure 4) potentially mediated through CD4
apy, anti-BTLA monotherapy, or combination therapy were + and CD8 + T cells (Figures 2, Figures 3). To determine whether
sorted for live CD45+ CD3+ CD8 + T cells and co-incubated CD4+ and CD8 + T cells are necessary for the observed survival
with dendritic cells and GL261-Luc2 tumor cell lysate benefit with combination therapy, we performed a depletion
(Figure 3a). Resultant supernatant with secreted IFN-γ was study (Figure 5). After confirming tumor burden on post-
then analyzed by ELISA. IFN-γ levels directly correlated with implantation day 7, mice were depleted of either CD4 + T cells
treatment, with significantly increased secretion of IFN-γ in or CD8 + T cells. After depletion of these T cell populations,
combination treatment compared to no treatment (P = .0033) mice were treated with anti-PD-1 plus anti-BTLA combination
and anti-BTLA monotherapy (P = .0032). There is a trend therapy. Depletion of CD4+ or CD8 + T cells was confirmed on
toward increased secretion of IFN-γ in combination treatment Day 14 (Supplemental Figure S6). There was no significant
compared to anti-PD-1 monotherapy (P = .0776) (Figure 3b). difference in survival between the control arm, CD4 + T cell
depletion plus combination therapy arm, and the CD8 + T cell
depletion plus combination therapy arm. Combination therapy
Combination treatment with anti-PD-1 and anti-BTLA without any T cell depletion had significantly greater median
results in improved overall long-term survival overall survival compared to control (P = .004), CD4 + T cell
depletion plus combination therapy (P = .036), and CD8 + T cell
Having confirmed BTLA expression on Tregs in GBM, we
depletion plus combination therapy (P = .046).
hypothesized that dual blockade with anti-PD-1 and anti-
BTLA antibodies would result in a synergistic survival benefit
due to targeted reversal of immunosuppression of CD8+ and
Discussion
CD4 + T cells. After ensuring tumor burden with GL261-Luc2
on post-implantation day 7, mice were treated with three doses Despite advances in immune checkpoint blockade for solid
of BTLA on days 7, 10, and 14 as well as anti-PD-1 on days 10, tumors, GBM has demonstrated resistance to anti-PD-1 ther­
12, and 14 per previous protocols19 (Figure 4a, b). Compared to apy with multiple mechanisms of immunosuppression.9 Of
anti-PD-1 monotherapy, combination treatment with dual note, beyond PD-1, TILs in GBM demonstrate upregulation
checkpoint blockade resulted in increased overall survival of additional inhibitory immune checkpoint molecules such as
(60% vs. 20%, P = .0003). There were no long-term survivors Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4),
with anti-BTLA monotherapy as well as no statistically signifi­ Indoleamine 2,3-dioxygenase 1 (IDO1), T-cell immunoglobu­
cant difference between control and anti-BTLA monotherapy lin and mucin-domain containing-3 (TIM-3), and
for median overall survival (P = .0954). On post-implantation Lymphocyte-activation gene-3 (LAG-3).17 As such, the utiliza­
day 60, long-term survivors and naïve control mice were tion of combination checkpoint therapy involving additional

Figure 3. In vitro co-culture assay for IFN-γ secretion with ELISA. (a) CD8 + T cells were isolated via FACS microfluidic sorting from untreated mice and mice treated
in vivo with anti-PD-1 monotherapy, anti-BTLA monotherapy, and combination anti-PD-1 and anti-BTLA. Cells were co-cultured with GL261 tumor cell lysate and CD11c
+ dendritic cells. (b) Plot of concentration of IFN-γ in supernatant of co-culture wells. All plots represent mean with standard error. Differences between two treatment
arms were analyzed via Student’s t test. (**** P < .0001, *** P < .001, ** P < .01, * P < .05).
e1956142-6 J. CHOI ET AL.

Figure 4. Survival schema and Kaplan-Meier curve. (a) Mice were implanted with 1.3e5 GL261-Luc2 cells with a stereotactic frame. On post-implantation day 7, mice
were assessed for presence of tumor by injecting them with 200 μl of 1 mg/ml D-luciferin and imaged with IVIS (In Vivo Imaging Systems), and subsequently randomly
assorted into four groups: control (no treatment), anti-BTLA monotherapy, anti-PD-1 monotherapy, and combination anti-BTLA and anti-PD-1 therapy. 5 mice in each
arm were saved for blood and brain harvest to undergo flow cytometric analysis. Remaining 8 mice in each arm underwent a survival study and were rechallenged
on day 60. (b) Treatment with anti-BTLA was on days 7, 10, and 14. Treatment with anti-PD-1 was on days 10, 12, and 14. (c) Kaplan-Meier curve demonstrating
differences in overall long-term survival with combination therapy. (**** P < .0001, *** P < .001, ** P < .01, * P < .05).

checkpoint blockade such as antibodies against CTLA-4, LAG- populations with anti-TIM-3 and anti-PD-1, respectively,
3, TIM-3, and IDO1 have been explored in both pre-clinical these preclinical studies show proof of principle of the syner­
and clinical investigations. gistic survival benefit that can result from targeting multiple
In the clinical setting, the precedent for success with com­ pathways of immunosuppression.
bination checkpoint therapy for brain tumors is most salient in In this study, we found a synergistic survival benefit accom­
treatment for metastatic melanoma, in which anti-CTLA-4 and panying combination therapy with anti-PD-1 and anti-BTLA
anti-PD-1 combination therapy was approved by the FDA in in a mouse glioma model, with increased 4–1BB expression
2016 as a first-line therapy and showed promising results of and IFN-γ secretion from CD4+ and CD8 + T cells. There was
improved progression-free survival (PFS) compared with anti- also a decreased proportion of infiltrating Tregs of the CD4
PD-1 therapy alone.20 There is currently an ongoing trial that is + TILs (Figures 2, Figures 4c). In tumor-bearing mice, combi­
studying antibodies against PD-1 and LAG-3 (NCT02658981), nation therapy demonstrated T cells with significantly
with several other combination studies bolstered by promising increased IFN-γ expression in both CD4+ and CD8 + T cell
preclinical findings.11,12,21In preclinical GBM murine models, populations that correlated with survival outcomes. This rever­
combination therapy involving antibodies against TIM-3 (anti- sal of immunosuppression when compared to control and
TIM-3), anti-PD-1, and stereotactic radiation resulted in 100% monotherapy arms in conjunction with a decreased proportion
overall survival.11 As the success of this mechanism is thought of Treg infiltration demonstrates that BTLA therapy may pos­
to be in part from targeting both myeloid and lymphocyte sibly involve a CD4-based mechanism, likely involving
ONCOIMMUNOLOGY e1956142-7

Figure 5. CD4+ and CD8 + T cell depletion Kaplan-Meier curve. Mice in the depletion arms underwent depletion of either CD4 + T cells or CD8 + T cells prior to
treatment with anti-BTLA and anti-PD-1 therapy. There was no significant difference in survival between the control arm, CD4 + T cell depletion arm, and CD8 + T cell
depletion arm. Combination therapy without any T cell depletion had significantly greater median overall survival compared to control (P = .004), CD4 + T cell depletion
plus combination therapy (P = .036), and CD8 + T cell depletion plus combination therapy (P = .046).

decreased polarization of CD4+ TILs toward the Treg pheno­ population over tumor progression from day 7 to day 21. Our
type in the TME, leading to a greater proportion of non-Treg data also showed that there was not a statistically significant
effector CD4+ TILs (Figure 2a, b, 3b). Furthermore, our deple­ difference in the infiltration of Tregs between anti-BTLA and
tion study (Figure 4) suggests that both CD4 + T cells and combination therapy arms. This has two possible implications:
CD8 + T cells are necessary for the synergistic survival benefit (1) the immune phenomenon affecting Treg infiltrating with
observed with anti-PD-1 and anti-BTLA combination therapy combination therapy is likely due to the inhibition of BTLA
(Figure 5). rather than PD-1 and (2) despite a similar decrease in Treg
CD4+ FoxP3+ Treg cells in the TME of GBM are infiltration, CD8+ IFN-γ expression as well as survival outcomes
thought to play an important role in immunosuppression. with anti-BTLA monotherapy failed to reach the same level as
Both preclinical and clinical studies have shown increased anti-PD-1 monotherapy or combination therapy (Figures 2b,
infiltration of Tregs within GBM,22–24 with data showing Figures 4). Interestingly, while the immunosuppressive role of
that the presence of these increased Treg cell fractions in Tregs have been well explored in both preclinical models and in
patients correlates with decreased effector T cell prolifera­ human GBM tissue, the impact of Tregs on overall survival and
tion and responsiveness.25 Several studies have since prognosis are less clear. An immunohistochemical analysis of
explored the potential survival impact of Treg based ther­ glioma tissue by Heimberger et al. with multivariate analysis for
apy in GBM,26 with anti-CD25 or anti-CTLA demonstrat­ the presence of Tregs on overall survival found that when
ing preclinical efficacy by targeting Tregs.22,27,28 However, accounting for confounding factors such as age and Karnofsky
these therapies have not yet successfully been translated performance score, the presence of FoxP3+ Tregs did not have
into the clinical setting.29 a significant prognostic impact.10 This may in part offer insights
Interestingly, our BTLA timepoint experiment (Figure 1) as to why anti-BTLA monotherapy may not confer as great
demonstrated differential expression of BTLA on tumor- a survival benefit, but in turn is an excellent candidate for
infiltrating CD4+ FoxP3+ Treg cells and CD4+ FoxP3- non- combination immunotherapy as targeting Tregs has been
Treg cells over time. BTLA expression remained constant over shown to impact immunosuppression in GBM.10
time on tumor-infiltrating Treg cells, however BTLA expression Additional lines of exploration that would be essential in
significantly increased over time on tumor-infiltrating non-Treg characterizing this avenue of treatment would be to delve into
cells. Indeed, there is a similar percentage of BTLA expression on further mechanisms involving BTLA, including understanding of
both tumor-infiltrating Treg cells and tumor-infiltrating non- involved essential trafficking molecules since our data suggests
Treg cells on day 7 (13.4% vs. 10.1% respectively). However, anti-BTLA decreases Treg infiltration into the brain. For example,
there is significantly higher BTLA expression on tumor- a notable target that has already been shown to be highly asso­
infiltrating non-Treg cells compared to tumor-infiltrating Treg ciated with Treg trafficking in GBM is C-C chemokine receptor
cells by day 21 (37.8% vs 20.0%). This data parallels data in type 4 (CCR4), which is highly expressed on Tregs with its cognate
human GBM patients showing PD-1 expression on TILs is antigen being produced by GBM.31 Furthermore, our supplemen­
increased in patients with GBM compared to control.30 tal data suggests BTLA and HVEM are also expressed on myeloid
Given that BTLA is an immune checkpoint, we posit that and B cells. In our study, we did not find a significant change in
tumor-infiltrating non-Treg cells become more suppressed over tumor-infiltrating CD11b+ cells, CD11c+ cells, or CD19 + B cells
time due to increased BTLA expression over the course of tumor with combination therapy compared to control. Despite this
progression. We believe there may not be BTLA-mediated sup­ result, It may be interesting to characterize in future studies
pression of tumor-infiltrating Treg cells given constant low whether anti-BTLA therapy affects the interaction between mye­
expression of the BTLA immune checkpoint on this cell loid or B cells and TILs.
e1956142-8 J. CHOI ET AL.

With any animal model research, consideration must be microenvironment. In this study, we study synergy between anti-BTLA
given to the degree to which the animal model accurately and anti-PD-1 immunotherapy and show that combination therapy sig­
mimics human disease. GBM is notorious for being difficult nificantly improves survival in a murine GBM model. Furthermore, we
demonstrate a direct effect of combination therapy on several immune cell
to replicate in vivo. Indeed, many therapeutic combinations populations including CD4+, CD8+, and regulatory T cells.
that work well in murine glioma models fail to translate to
clinical trials in human GBM.6,19,32 In this project, all
experiments were conducted using the GL261-Luc2 cell Disclosure statement
line. The GL261 cell line is one of the most well-studied
The authors do not have any relevant conflicts of interest associated with
glioma models in the field of GBM immunotherapy.33 The this manuscript.
primary benefit of this cell line is that it is well character­
ized, can be easily implanted into immunocompetent
C57BL/6 mice given that it is syngeneic, and effectively Funding
recapitulates many of the characteristics of human
This research was funded from a gift fund by private donors Conflict of
GBM.34,35 The Luc2 tag allows for tumor visualization on Interest: Michael Lim (Funding from Arbor Pharmaceuticals, Accuray,
IVIS, and indeed many recent studies evaluating checkpoint BMS, Novartis; Consultant: BMS, Merck, SQZ Biotechnologies, Tocagen,
inhibition therapy in GBM have used the GL261-Luc2 VBI; Patents: Combining Focused Radiation and Immunotherapy,
model.11,19,32,36 The primary disadvantage of this model is Combining Local Chemotherapy and Immunotherapy)
that it does not fully recapitulate the significant intra-
tumoral and inter-tumoral heterogeneity of GBM.
Furthermore, the GL261-Luc2 model is a moderately ORCID
immunogenic model whereas GBM is significantly Dimitrios Mathios http://orcid.org/0000-0002-0792-6880
immunoresistant.35 It is important to bear in mind the Michael Lim http://orcid.org/0000-0003-0523-3076
strengths and limitations of the preclinical model utilized
to study GBM in the preclinical setting, and additional
experiments should be conducted in other GBM cell lines
Data availability statement:
to improve generalizability of these results. The data that support the findings of this study are available from the
The appeal of BTLA as a target for immunotherapy in corresponding author upon reasonable request.
GBM comes from its multimodal effect on regulatory
T cells and CD8 + T cells. The dramatic changes in Treg
Ethics statement:
representation as well as the synergistic survival benefit
seen in murine in vivo models demonstrate a treatment This study protocol was approved by the Johns Hopkins Institutional
schema that can target multiple immunosuppressive cells Animal Care and Use Committee. This study did not involve any
when used in combination with antibodies involved in the human patients.
well-established PD-1 pathway. With immune checkpoint
therapy continuing to gain traction in the clinical setting, References
new targets that synergize survival outcomes are of interest.
1. Ostrom QT, Gittleman H, Liao P, Vecchione-Koval T, Wolinsky Y,
Our preclinical study involving antibodies against PD-1 and Kruchko C, Barnholtz-Sloan JS. CBTRUS statistical report: pri­
BTLA offer proof of principle for the efficacy of this com­ mary brain and other central nervous system tumors diagnosed
bination therapy and offers the first exploration of targeting in the United States in 2010-2014. Neuro Oncol. 2017 Nov;19(5):
BTLA in GBM in a mouse model. v1–88. doi:10.1093/neuonc/nox158.
2. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B,
Taphoorn MJB, Belanger K, Brandes AA, Marosi C, Bogdahn U,
Abbreviations et al. Radiotherapy plus concomitant and adjuvant temozolomide
for glioblastoma. N Engl J Med. 2005 Mar;352(10):987–996.
Band T lymphocyte attenuator (BTLA); Bristol Myers Squibb (BMS); doi:10.1056/NEJMoa043330.
C-C chemokine receptor type 4 (CCR4); Cytotoxic T-lymphocyte- 3. Di Carlo DT, Cagnazzo F, Benedetto N, Morganti R, Perrini P.
associated protein 4 (CTLA-4); Fluorescence minus one (FMO); Multiple high-grade gliomas: epidemiology, management, and out­
Glioblastoma (GBM); Herpes virus entry mediator (HVEM); immune come. A systematic review and meta-analysis. Vol. 42,
checkpoint blockade (ICB); Indoleamine 2,3-dioxygenase 1 (IDO1); Neurosurgical Review. Springer Verlag; 2019. p. 263–275.
Institutional Animal Care and Use Committee (IACUC); intra- 4. Paz-Ares L, Luft A, Vicente D, Tafreshi A, Gümüş M, Mazières J,
peritoneal (i.p.); IVIS (In Vivo Imaging System); Lymphocyte-activation et al. Pembrolizumab plus chemotherapy for squamous non–
gene-3 (LAG-3); median overall survival (mOS); monoclonal antibodies small-cell lung cancer. N Engl J Med. 2018 Nov;379
(mAbs); non-small cell lung cancer (NSCLC); phosphate-buffered saline (21):2040–2051. doi:10.1056/NEJMoa1810865.
(PBS); programmed cell death protein-1 (anti-PD-1); progression-free 5. Paz-Ares L, Horn L, Borghaei H, Spigel DR, Steins M, Ready N,
survival (PFS); regulatory T cells (Tregs); T-cell immunoglobulin and et al. Phase III, randomized trial (CheckMate 057) of nivolumab
mucin-domain containing-3 (TIM-3); tumor infiltrating lymphocytes (NIVO) versus docetaxel (DOC) in advanced non-squamous cell
(TILs); tumor microenvironment (TME); (non-SQ) non-small cell lung cancer (NSCLC). J Clin Oncol. 2015
Jun;33(18_suppl):LBA109–LBA109. doi:10.1200/jco.2015.33.18_­
suppl.lba109.
Novelty and Impact: 6. Omuro A, Vlahovic G, Lim M, Sahebjam S, Baehring J,
Cloughesy T, Voloschin A, Ramkissoon SH, Ligon KL, Latek R,
BTLA is an immune checkpoint thought to downregulate the immune et al. Nivolumab with or without ipilimumab in patients with
system. BTLA has been shown to be upregulated in the GBM tumor recurrent glioblastoma: results from exploratory phase i cohorts
ONCOIMMUNOLOGY e1956142-9

of CheckMate 143. Neuro Oncol. 2018;20(5):674–686. doi:10.1093/ 22. El AA, Lesniak MS. An increase in CD4+CD25+FOXP3+ regula­
neuonc/nox208. tory T cells in tumor-infiltrating lymphocytes of human glioblas­
7. Lim M, Xia Y, Bettegowda C, Weller M. Current state of immu­ toma multiforme1. Neuro Oncol. 2006 Jul;8(3):234–243.
notherapy for glioblastoma. Vol. 15, nature reviews clinical oncol­ doi:10.1215/15228517-2006-006.
ogy. Nature Publishing Group; 2018. p. 422–442. 23. Sugihara AQ, Rolle CE, Lesniak MS. Regulatory T cells actively
8. Razavi SM, Lee KE, Jin BE, Aujla PS, Gholamin S, Li G. Immune infiltrate metastatic brain tumors. Int J Oncol. 2009;34:1533–1540.
evasion strategies of glioblastoma. Vol. 3, frontiers in surgery. 24. Kennedy BC, Maier LM, D’Amico R, Mandigo CE, Fontana EJ,
Frontiers Media S.A.; 2016. p. 11. Waziri A, Assanah MC, Canoll P, Anderson RC, Anderson DE,
9. Jackson CM, Choi J, Lim M. Mechanisms of immunotherapy et al. Dynamics of central and peripheral immunomodulation in
resistance: lessons from glioblastoma. Vol. 20, nature immunology. a murine glioma model. BMC Immunol. 2009 Feb;10(1):11.
Nature Publishing Group; 2019. p. 1100–1109. doi:10.1186/1471-2172-10-11.
10. Heimberger AB, Abou-Ghazal M, Reina-Ortiz C, Yang DS, Sun W, 25. Fecci PE, Mitchell DA, Whitesides JF, Xie W, Friedman AH,
Qiao W, Hiraoka N, Fuller GN. Incidence and prognostic impact of Archer GE, Herndon JE, Bigner DD, Dranoff G, Sampson JH,
FoxP3+regulatory T cells in human gliomas. Clin Cancer Res. 2008 et al. Increased regulatory T-cell fraction amidst a diminished
Aug;14(16):5166–5172. doi:10.1158/1078-0432.CCR-08-0320. CD4 compartment explains cellular immune defects in patients
11. Kim JE, Patel MA, Mangraviti A, Kim ES, Theodros D, Velarde E, with malignant glioma. Cancer Res. 2006 Mar;66(6):3294–3302.
Liu A, Sankey EW, Tam A, Xu H, et al. Combination therapy with doi:10.1158/0008-5472.CAN-05-3773.
anti-PD-1, anti-TIM-3, and focal radiation results in regression of 26. Lohr J, Ratliff T, Huppertz A, Ge Y, Dictus C, Ahmadi R, Grau S,
murine gliomas. Clin Cancer Res. 2017 Jan;23(1):124–136. Hiraoka N, Eckstein V, Ecker RC, et al. Effector T-cell infiltration
doi:10.1158/1078-0432.CCR-15-1535. positively impacts survival of glioblastoma patients and is impaired
12. Harris-Bookman S, Mathios D, Martin AM, Xia Y, Kim E, Xu H, by tumor-derived TGF-β. Clin Cancer Res. 2011 Jul;17
Belcaid Z, Polanczyk M, Barberi T, Theodros D, et al. Expression of (13):4296–4308. doi:10.1158/1078-0432.CCR-10-2557.
LAG-3 and efficacy of combination treatment with anti-LAG-3 and 27. Fecci PE, Sweeney AE, Grossi PM, Nair SK, Learn CA,
anti-PD-1 monoclonal antibodies in glioblastoma. Int J Cancer. Mitchell DA, Cui X, Cummings TJ, Bigner DD, Gilboa E, et al.
2018 Dec;143(12):3201–3208. doi:10.1002/ijc.31661. Systemic anti-CD25 monoclonal antibody administration safely
13. Watanabe N, Gavrieli M, Sedy JR, Yang J, Fallarino F, Loftin SK, enhances immunity in murine glioma without eliminating regula­
Hurchla MA, Zimmerman N, Sim J, Zang X, et al. BTLA is tory T cells. Clin Cancer Res. 2006;12(14 Pt 1):4294–4305.
a lymphocyte inhibitory receptor with similarities to CTLA-4 and doi:10.1158/1078-0432.CCR-06-0053.
PD-1. Nat Immunol. 2003 Jul;4(7):670–679. doi:10.1038/ni944. 28. Selby MJ, Engelhardt JJ, Quigley M, Henning KA, Chen T,
14. Derré L, Rivals JP, Jandus C, Pastor S, Rimoldi D, Romero P, Srinivasan M, Korman AJ. Anti-CTLA-4 antibodies of IgG2a iso­
Michielin O, Olive D, Speiser DE. BTLA mediates inhibition of type enhance antitumor activity through reduction of intratumoral
human tumor-specific CD8+ T cells that can be partially reversed regulatory T cells. Cancer Immunol Res. 2013 Jul;1(1):32–42.
by vaccination. J Clin Invest. 2010 Jan;120(1):157–167. doi:10.1158/2326-6066.CIR-13-0013.
doi:10.1172/JCI40070. 29. Lim M;, Weller M;, Chiocca EA Current State of Immune-Based
15. Lan X, Li S, Gao H, Nanding A, Quan L, Yang C, Ding S, Xue Y. Therapies for Glioblastoma. 2016;
Increased BTLA and HVEM in gastric cancer are associated with 30. Davidson TB, Lee A, Hsu M, Sedighim S, Orpilla J, Treger J,
progression and poor prognosis. Onco Targets Ther. 2017 Mastall M, Roesch S, Rapp C, Galvez M. et al. Expression of
Feb;10:919–926. doi:10.2147/OTT.S128825. PD-1 by T Cells in Malignant Glioma Patients Reflects
16. Liu J, Li J, He M, Zhang GL, Zhao Q. Distinct changes of BTLA and Exhaustion and Activation. Clin Cancer Res. Internet]. 2019 Mar
HVEM expressions in circulating CD4+ and CD8+ T cells in hepa­ 15;25(6):1913 LP– 1922. Available from. ;:. 10.1158/1078-0432.
tocellular carcinoma patients. J Immunol Res. 2018;2018: 4561571. CCR-18-1176
17. Woroniecka K, Chongsathidkiet P, Rhodin K, Kemeny H, 31. Jordan JT, Sun W, Hussain SF, DeAngulo G, Prabhu SS,
Dechant C, Farber SH, Elsamadicy AA, Cui X, Koyama S, Heimberger AB. Preferential migration of regulatory T cells
Jackson C. et al. T-cell exhaustion signatures vary with tumor mediated by glioma-secreted chemokines can be blocked with
type and are severe in glioblastoma. Clin Cancer Res. Internet]. chemotherapy. Cancer Immunol Immunother. 2008 May;57
2018 Sep 1;24(17):4175LP–4186. Available from. ;:. 10.1158/1078- (1):123–131. doi:10.1007/s00262-007-0336-x.
0432.CCR-17-1846 32. Reardon DA, Gokhale PC, Klein SR, Ligon KL, Rodig SJ,
18. Han MZ, Wang S, Zhao WB, Ni SL, Yang N, Kong Y, Huang B, Ramkissoon SH, Jones KL, Conway AS, Liao X, Zhou J, et al.
Chen A-J, Li X-G, Wang J, et al. Immune checkpoint molecule Glioblastoma eradication following immune checkpoint block­
herpes virus entry mediator is overexpressed and associated with ade in an orthotopic, immunocompetent model. Cancer
poor prognosis in human glioblastoma. EBioMedicine. 2019 Immunol Res. 2016 Feb;4(2):124–135. doi:10.1158/2326-6066.
May;43:159–170. doi:10.1016/j.ebiom.2019.04.002. CIR-15-0151.
19. Zeng J, See AP, Phallen J, Jackson CM, Belcaid Z, Ruzevick J, 33. Maes W, Van Gool SW. Experimental immunotherapy for malig­
Durham N, Meyer C, Harris TJ, Albesiano E, et al. Anti-PD-1 nant glioma: lessons from two decades of research in the GL261
blockade and stereotactic radiation produce long-term survival in model. Cancer Immunol Immunother. 2011 Feb;60(2):153–160.
mice with intracranial gliomas. Int J Radiat Oncol Biol Phys. 2013 doi:10.1007/s00262-010-0946-6.
Jun;86(2):343–349. doi:10.1016/j.ijrobp.2012.12.025. 34. Lenting K, Verhaak R, Ter Laan M, Wesseling P, Leenders W.
20. Hodi FS, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Glioma: experimental models and reality. Acta Neuropathol.
Cowey CL, Lao CD, Schadendorf D, Wagstaff J, Dummer R, et al. 2017 Feb;133(2):263–282. doi:10.1007/s00401-017-1671-4.
Nivolumab plus ipilimumab or nivolumab alone versus ipilimu­ 35. Oh T, Fakurnejad S, Sayegh ET, Clark AJ, Ivan ME, Sun MZ et al.
mab alone in advanced melanoma (CheckMate 067): 4-year out­ Immunocompetent murine models for the study of glioblastoma
comes of a multicentre, randomised, phase 3 trial. Lancet Oncol. immunotherapy J Transl Med 2014;12:107Internet]. ;(1):. Available
2018 Nov;19(11):1480–1492. doi:10.1016/S1470-2045(18)30700-9. from 10.1186/1479-5876-12-107
21. Wainwright DA, Chang AL, Dey M, Balyasnikova IV, Kim CK, 36. Mathios D, Kim JE, Mangraviti A, Phallen J, Park C-K,
Tobias A, Cheng Y, Kim JW, Qiao J, Zhang L, et al. Durable Jackson CM. et al. Anti-PD-1 antitumor immunity is enhanced
therapeutic efficacy utilizing combinatorial blockade against IDO, by local and abrogated by systemic chemotherapy in GBM. Sci
CTLA-4, and PD-L1 in mice with brain tumors. Clin Cancer Res. Transl Med. Internet]. 2016 Dec 21;8(370):370ra180–370ra180.
2014 Oct;20(20):5290–5301. doi:10.1158/1078-0432.CCR-14-0514. Available from. ;:. 10.1126/scitranslmed.aag2942

You might also like